Dondorp 2010 Artesunate versus quinine in the treatment of severe falciparum malaria

11
Articles www.thelancet.com Vol 376 November 13, 2010 1647 Lancet 2010; 376: 1647–57 Published Online November 8, 2010 DOI:10.1016/S0140- 6736(10)61924-1 This online publication has been corrected. The corrected version first appeared at thelancet.com on January 7, 2011 See Comment page 1621 *Members listed at end of paper. Hospital Central da Beira, Beira, Mozambique (E Gomes MD, A Seni MD, K D Chhaganlal MD); MRC laboratories, Banjul, The Gambia (K Bojang FRCP, R Olaosebikan FWACP, N Anunobi FMCpaed); Komfo Anokye Hospital, Kumasi, Ghana (Prof T Agbenyega PhD, S Blay Nguah FWACPCH, J Evans MRCPCH); Kilifi District General Hospital, Kilifi, Kenya (Prof K Maitland PhD, E Kivaya MSc); Magunga District Hospital, NIMR-Korogwe Research Laboratory, Tanga, Tanzania (S Gesase MD, C Kahabuka MD); Teule Designated District Hospital, Muheza, Tanzania (B Nadjm MD, J Deen MD); NIMR-Amani Centre, Tanga, Tanzania (G Mtove, MD); Malaria Control Program, Ministry of Health, Kigali, Rwanda (C Karema MD, N Umulisa MD, A Uwimana MD); University of Ilorin Teaching Hospital, Ilorin, Nigeria (O A Mokuolu FWACP, O T Adedoyin FWACP, Prof W B R Johnson FWACP); Mbarara University of Science and Technology and Epicentre Research Base, Mbarara, Uganda (J Mwanga-Amumpaire MD, M Nansumba MD); Kinshasa School of Public Health— Kingasani Research Centre, Kinshasa, Democratic Republic of the Congo Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial Arjen M Dondorp, Caterina I Fanello, Ilse C E Hendriksen, Ermelinda Gomes, Amir Seni, Kajal D Chhaganlal, Kalifa Bojang, Rasaq Olaosebikan, Nkechinyere Anunobi, Kathryn Maitland, Esther Kivaya, Tsiri Agbenyega, Samuel Blay Nguah, Jennifer Evans, Samwel Gesase, Catherine Kahabuka, George Mtove, Behzad Nadjm, Jacqueline Deen, Juliet Mwanga-Amumpaire, Margaret Nansumba, Corine Karema, Noella Umulisa, Aline Uwimana, Olugbenga A Mokuolu, Olanrewaju T Adedoyin, Wahab B R Johnson, Antoinette K Tshefu, Marie A Onyamboko, Tharisara Sakulthaew, Wirichada Pan Ngum, Kamolrat Silamut, Kasia Stepniewska, Charles J Woodrow, Delia Bethell, Bridget Wills, Martina Oneko, Tim E Peto, Lorenz von Seidlein, Nicholas P J Day, Nicholas J White, for the AQUAMAT group* Summary Background Severe malaria is a major cause of childhood death and often the main reason for paediatric hospital admission in sub-Saharan Africa. Quinine is still the established treatment of choice, although evidence from Asia suggests that artesunate is associated with a lower mortality. We compared parenteral treatment with either artesunate or quinine in African children with severe malaria. Methods This open-label, randomised trial was undertaken in 11 centres in nine African countries. Children (<15 years) with severe falciparum malaria were randomly assigned to parenteral artesunate or parenteral quinine. Randomisation was in blocks of 20, with study numbers corresponding to treatment allocations kept inside opaque sealed paper envelopes. The trial was open label at each site, and none of the investigators or trialists, apart from for the trial statistician, had access to the summaries of treatment allocations. The primary outcome measure was in-hospital mortality, analysed by intention to treat. This trial is registered, number ISRCTN50258054. Findings 5425 children were enrolled; 2712 were assigned to artesunate and 2713 to quinine. All patients were analysed for the primary outcome. 230 (8·5%) patients assigned to artesunate treatment died compared with 297 (10·9%) assigned to quinine treatment (odds ratio [OR] stratified for study site 0·75, 95% CI 0·63–0·90; relative reduction 22·5%, 95% CI 8·1–36·9; p=0·0022). Incidence of neurological sequelae did not differ significantly between groups, but the development of coma (65/1832 [3·5%] with artesunate vs 91/1768 [5·1%] with quinine; OR 0·69 95% CI 0·49–0·95; p=0·0231), convulsions (224/2712 [8·3%] vs 273/2713 [10·1%]; OR 0·80, 0·66–0·97; p=0·0199), and deterioration of the coma score (166/2712 [6·1%] vs 208/2713 [7·7%]; OR 0·78, 0·64–0·97; p=0·0245) were all significantly less frequent in artesunate recipients than in quinine recipients. Post-treatment hypoglycaemia was also less frequent in patients assigned to artesunate than in those assigned to quinine (48/2712 [1·8%] vs 75/2713 [2·8%]; OR 0·63, 0·43–0·91; p=0·0134). Artesunate was well tolerated, with no serious drug-related adverse effects. Interpretation Artesunate substantially reduces mortality in African children with severe malaria. These data, together with a meta-analysis of all trials comparing artesunate and quinine, strongly suggest that parenteral artesunate should replace quinine as the treatment of choice for severe falciparum malaria worldwide. Funding The Wellcome Trust. Introduction Falciparum malaria is a major contributor to child mortality in Africa and one of the main causes of paediatric hospital admission across sub-Saharan Africa. Many deaths occur in or near the home, but for children who are admitted to hospital with severe malaria and receive parenteral antimalarial treatment, about one in six will die. 1,2 From the time of its introduction to European medicine in the 1630s until the deployment of parenteral chloroquine in the 1950s, quinine was the mainstay of severe malaria treatment. Resistance to chloroquine emerged in southeast Asia and then spread to Africa at the end of the 1970s. Quinine then resumed its primary role in the treatment of severe malaria. Parenteral quinine has a narrow therapeutic ratio. 1–3 Intravenous quinine administration needs a constant rate infusion with dosing three times a day. Intramuscular administration is painful, and can cause sterile abscesses and predispose to lethal tetanus. 4 Although blindness and deafness may follow self poisoning, these side-effects are rare in severe malaria; however, quinine-induced hyperinsulinaemic hypoglycaemia is a particular problem in patient manage- ment, especially in pregnant women. 1,2,5 The primacy of quinine in the treatment of severe malaria has been challenged by the introduction of artemisinin derivatives. The first comparative clinical trials were done with intramuscular artemether—a lipophilic derivative of dihydroartemisinin. Artemether proved safer

Transcript of Dondorp 2010 Artesunate versus quinine in the treatment of severe falciparum malaria

Articles

www.thelancet.com Vol 376 November 13, 2010 1647

Lancet 2010; 376: 1647–57

Published OnlineNovember 8, 2010DOI:10.1016/S0140-6736(10)61924-1

This online publication has been corrected. The corrected version fi rst appeared at thelancet.com on January 7, 2011

See Comment page 1621

*Members listed at end of paper.

Hospital Central da Beira, Beira, Mozambique (E Gomes MD, A Seni MD, K D Chhaganlal MD); MRC laboratories, Banjul, The Gambia (K Bojang FRCP, R Olaosebikan FWACP, N Anunobi FMCpaed); Komfo Anokye Hospital, Kumasi, Ghana (Prof T Agbenyega PhD, S Blay Nguah FWACPCH, J Evans MRCPCH); Kilifi District General Hospital, Kilifi , Kenya (Prof K Maitland PhD, E Kivaya MSc); Magunga District Hospital, NIMR-Korogwe Research Laboratory, Tanga, Tanzania (S Gesase MD, C Kahabuka MD); Teule Designated District Hospital, Muheza, Tanzania (B Nadjm MD, J Deen MD); NIMR-Amani Centre, Tanga, Tanzania (G Mtove, MD); Malaria Control Program, Ministry of Health, Kigali, Rwanda (C Karema MD, N Umulisa MD, A Uwimana MD); University of Ilorin Teaching Hospital, Ilorin, Nigeria (O A Mokuolu FWACP, O T Adedoyin FWACP, Prof W B R Johnson FWACP); Mbarara University of Science and Technology and Epicentre Research Base, Mbarara, Uganda (J Mwanga-Amumpaire MD, M Nansumba MD); Kinshasa School of Public Health—Kingasani Research Centre, Kinshasa, Democratic Republic of the Congo

Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trialArjen M Dondorp, Caterina I Fanello, Ilse C E Hendriksen, Ermelinda Gomes, Amir Seni, Kajal D Chhaganlal, Kalifa Bojang, Rasaq Olaosebikan, Nkechinyere Anunobi, Kathryn Maitland, Esther Kivaya, Tsiri Agbenyega, Samuel Blay Nguah, Jennifer Evans, Samwel Gesase, Catherine Kahabuka, George Mtove, Behzad Nadjm, Jacqueline Deen, Juliet Mwanga-Amumpaire, Margaret Nansumba, Corine Karema, Noella Umulisa, Aline Uwimana, Olugbenga A Mokuolu, Olanrewaju T Adedoyin, Wahab B R Johnson, Antoinette K Tshefu, Marie A Onyamboko, Tharisara Sakulthaew, Wirichada Pan Ngum, Kamolrat Silamut, Kasia Stepniewska, Charles J Woodrow, Delia Bethell, Bridget Wills, Martina Oneko, Tim E Peto, Lorenz von Seidlein, Nicholas P J Day, Nicholas J White, for the AQUAMAT group*

SummaryBackground Severe malaria is a major cause of childhood death and often the main reason for paediatric hospital admission in sub-Saharan Africa. Quinine is still the established treatment of choice, although evidence from Asia suggests that artesunate is associated with a lower mortality. We compared parenteral treatment with either artesunate or quinine in African children with severe malaria.

Methods This open-label, randomised trial was undertaken in 11 centres in nine African countries. Children (<15 years) with severe falciparum malaria were randomly assigned to parenteral artesunate or parenteral quinine. Randomisation was in blocks of 20, with study numbers corresponding to treatment allocations kept inside opaque sealed paper envelopes. The trial was open label at each site, and none of the investigators or trialists, apart from for the trial statistician, had access to the summaries of treatment allocations. The primary outcome measure was in-hospital mortality, analysed by intention to treat. This trial is registered, number ISRCTN50258054.

Findings 5425 children were enrolled; 2712 were assigned to artesunate and 2713 to quinine. All patients were analysed for the primary outcome. 230 (8·5%) patients assigned to artesunate treatment died compared with 297 (10·9%) assigned to quinine treatment (odds ratio [OR] stratifi ed for study site 0·75, 95% CI 0·63–0·90; relative reduction 22·5%, 95% CI 8·1–36·9; p=0·0022). Incidence of neurological sequelae did not diff er signifi cantly between groups, but the development of coma (65/1832 [3·5%] with artesunate vs 91/1768 [5·1%] with quinine; OR 0·69 95% CI 0·49–0·95; p=0·0231), convulsions (224/2712 [8·3%] vs 273/2713 [10·1%]; OR 0·80, 0·66–0·97; p=0·0199), and deterioration of the coma score (166/2712 [6·1%] vs 208/2713 [7·7%]; OR 0·78, 0·64–0·97; p=0·0245) were all signifi cantly less frequent in artesunate recipients than in quinine recipients. Post-treatment hypoglycaemia was also less frequent in patients assigned to artesunate than in those assigned to quinine (48/2712 [1·8%] vs 75/2713 [2·8%]; OR 0·63, 0·43–0·91; p=0·0134). Artesunate was well tolerated, with no serious drug-related adverse eff ects.

Interpretation Artesunate substantially reduces mortality in African children with severe malaria. These data, together with a meta-analysis of all trials comparing artesunate and quinine, strongly suggest that parenteral artesunate should replace quinine as the treatment of choice for severe falciparum malaria worldwide.

Funding The Wellcome Trust.

IntroductionFalciparum malaria is a major contributor to child mortality in Africa and one of the main causes of paediatric hospital admission across sub-Saharan Africa. Many deaths occur in or near the home, but for children who are admitted to hospital with severe malaria and receive parenteral antimalarial treatment, about one in six will die.1,2 From the time of its introduction to European medicine in the 1630s until the deployment of parenteral chloroquine in the 1950s, quinine was the mainstay of severe malaria treatment. Resistance to chloroquine emerged in southeast Asia and then spread to Africa at the end of the 1970s. Quinine then resumed its primary role in the treatment of severe malaria. Parenteral quinine

has a narrow therapeutic ratio.1–3 Intravenous quinine administration needs a constant rate infusion with dosing three times a day. Intramuscular administration is painful, and can cause sterile abscesses and predispose to lethal tetanus.4 Although blindness and deafness may follow self poisoning, these side-eff ects are rare in severe malaria; however, quinine-induced hyperinsulinaemic hypoglycaemia is a particular problem in patient manage-ment, especially in pregnant women.1,2,5

The primacy of quinine in the treatment of severe malaria has been challenged by the introduction of artemisinin derivatives. The fi rst comparative clinical trials were done with intramuscular artemether—a lipophilic derivative of dihydroartemisinin. Artemether proved safer

Articles

1648 www.thelancet.com Vol 376 November 13, 2010

and easier to use than quinine, but did not improve overall survival in an individual patient data meta-analysis6 of 1919 randomised patients. In a prospectively defi ned subgroup analysis, artemether reduced mortality signifi cantly in southeast Asian adults, but not in African

children. A large multicentre randomised trial (South East Asian Quinine Artesunate Malaria Trial [SEAQUAMAT]),7 which compared intra venous artesunate with quinine in Asian patients (mainly adults) with severe malaria was then undertaken. It was stopped after enrolment of 1461 patients because of a substantial survival benefi t in favour of artesunate. In a meta-analysis of the SEAQUAMAT study and earlier, smaller randomised trials,7 artesunate reduced the mortality of severe malaria in Asian patients from 23·1% to 14·2%, a relative reduction of 38·6%. The treatment was also highly cost eff ective.8 In 2006, WHO changed its guidelines to recommend artesunate for severe malaria in adults.9

The treatment eff ect in the SEAQUAMAT trial was similar in adults and the 202 children enrolled, but perceived diff erences in the natural history and drug susceptibility of severe falciparum malaria in African children compared with Asian patients left uncertainty about the optimum treatment for this important patient group. Expert opinion regarded parenteral quinine as a satisfactory treatment, and research priorities focused on improving other aspects of the care of the sick child. Nowadays quinine remains by far the most widely used treatment of severe malaria in Africa. We undertook a large multicentre randomised trial (African Quinine Artesunate Malaria Trial [AQUAMAT]) that compared parenteral treatment with either artesunate or quinine in African children with severe malaria.

MethodsStudy designThis was a multicentre, open-label trial in children admitted to hospital with severe malaria, undertaken between Oct 3, 2005, and July 14, 2010. 11 centres in nine countries (Mozambique, The Gambia, Ghana, Kenya, Tanzania, Nigeria, Uganda, Rwanda, and Democratic Republic of the Congo) in Africa partici pated. The study was coordinated by the Mahidol-Oxford Tropical Medicine Research Unit in Bangkok, Thailand, which provided logistic support and data management. All clinicians were familiarised with the severe malaria criteria for enrolment (panel 1). Most children were managed on general paedi-atric wards. Children younger than 15 years were included if they had a positive rapid diagnostic test for Plasmodium falciparum lac tate dehydrogenase (Optimal, Diamed, Cressier, Switzer land) and, in the admitting physician’s clinical opinion, they had severe malaria, and they or their attendant relative or guardian gave fully informed written consent. Patients were not included if there was a convincing history of full treatment with parenteral quin-ine or an artemisinin derivative for more than 24 h before admission. Age criteria varied slightly between sites at the request of the respective ethics review boards (webappendix p 13). In Mozambique, adults were also studied but were analysed separately and will be reported elsewhere.

The trial protocol was reviewed and approved by each site’s ethics review board, and by the Oxford Tropical

(Prof A K Tshefu MD, M A Onyamboko MB); Mahidol

Oxford Tropical Medicine Research Unit (MORU), Faculty

of Tropical Medicine, Mahidol

University, Bangkok, Thailand (A M Dondorp MD,

C I Fanello PhD, I C E Hendriksen MD,

T Sakulthaew BNS, W Pan Ngum PhD, K Silamut PhD,

K Stepniewska PhD, C J Woodrow MRCP, Prof N P J Day FRCP,

Prof N J White FRS); Nuffi eld Department of Clinical

Medicine, University of Oxford, Oxford, UK (D Bethell MRCPCH,

Prof T E Peto FRCP); Menzies School of Health Research,

Casuarina, NT, Australia (L von Seidlein PhD); Oxford University Clinical Research

Unit, Ho Chi Minh City, Vietnam (B Wills FRCPCH); and KEMRI—

CDC Kisumu, Kisumu Kenya (M Oneko MD)

Panel 1: Modifi ed criteria for severe falciparum malaria

At least one of:• Plasma base excess less than –3·3 mmol/L• Glasgow coma scale less than 11 of 15, or Blantyre coma

scale less than 3 of 5 in preverbal children• Haemoglobin less than 50 g/L and parasitaemia greater

than 100 000 parasites per μL• Blood urea greater than 10 mmol/L• Compensated shock (capillary refi ll ≥3 s or temperature

gradient on legs, but no hypotension)• Decompensated shock; systolic blood pressure less than

70 mm Hg and cool peripheries• Asexual parasitaemia more than 10%• Visible jaundice and more than 100 000 parasites per μL• Plasma glucose less than 3 mmol/L• Respiratory distress, defi ned as costal indrawing, use of

accessory muscles, nasal alar fl aring, deep breathing, or severe tachypnoea

Figure 1: Trial profi le*663 in Beira, 442 in Kilifi , 436 in Kumasi, 921 in Muheza, 540 in Korogwe, 502 in Banjul, 450 in Ilorin, 386 in Rwanda, 663 in Mbarara, and 422 in Kinshasa. †Two patients also had other criteria.

11 148 assessed for eligibility

5425 enrolled*

2712 randomised to artesunate 2713 randomised to quinine

2712 analysed by intention to treat 2713 analysed by intention to treat

149 excluded from per-protocol analysis17 blood smear negative†

112 missing slides6 died before receiving drug7 missed early treatment dose(s)7 unknown number of treatment doses2 withdrawn consent

2563 analysed per protocol 2552 analysed per protocol

161 excluded from per-protocol analysis25 blood-smear negative99 missing slides22 died before receiving drug

4 missed early treatment dose(s)8 unknown number of treatment doses3 did not fulfil entry criteria

5723 excluded223 insufficient clinical severity criteria

4910 negative or invalid rapid diagnostic test180 parenteral antimalarial treatment >24 h316 refused consent

23 died before enrolment25 below age or weight criteria46 other reasons

For more about AQUAMAT see http://www.tropmedres.ac/

aquamat

Articles

www.thelancet.com Vol 376 November 13, 2010 1649

Research Ethics Committee. Permission to investigate HIV status was obtained only from some ethics review boards.

Randomisation and maskingEligible patients were randomly assigned to treatment with either intravenous or intramuscular artesunate or quinine. Each centre had a policy of using one route of administration. Randomisation was done by people unrelated to the study and provided to the study sites in blocks of 20. Study numbers were kept inside opaque sealed paper envelopes. After full informed written consent was obtained, the next envelope, which contained a unique study box number, was opened by the study physician or nurse. Then the corresponding numbered sealed box was opened. This box contained the study drug, case record

form (labelled with the unique study number), and all disposables needed for drug administration and blood sampling. Treatment was started immediately. All other

Quinine(N=2713)

Artesunate(N=2712)

Female sex 1295 (48%) 1315 (48%)

Age (years) 2·9 (1·7–4·3) 2·8 (1·6–4·2)

Fever before enrolment (days) 3 (2–4) 3 (2–4)

Coma before enrolment (h) 5·0 (2·5–10) 5·0 (2·0–9·5)

Pretreatment with antimalarials

None 1270 (47%) 1281 (47%)

Ineff ective* 371 (14%) 387 (15%)

Eff ective* 959 (37%) 938 (36%)

Complications on admission

Coma† 945 (35%) 880 (32%)

Convulsions 879 (32%) 811 (30%)

Jaundice 59 (2%) 55 (2%)

Severe anaemia (haemoglobin <50 g/L)

692 (29%) 737 (30%)

Shock 339 (12%) 323 (12%)

Decompensated shock 88 (35%) 90 (39%)

Severe acidosis (BE <–8 mmol/L) 975 (43%) 1009 (44%)

Hypoglycaemia (<3 mmol/L) 278 (10%) 277 (10%)

Respiratory distress‡ 428 (16%) 439 (16%)

Severe prostration§ 1668 (61%) 1683 (62%)

Blackwater fever 116 (4%) 121 (4%)

Hyperparasitaemia (>10%) 573 (24%) 584 (25%)

Clinical examination

Weight (kg) 12·6 (4·6) 12·4 (4·8)

Temperature (°C) 38·0 (1·1) 38·0 (1·1)

Blood pressure (mm Hg)

Systolic 95 (16) 95 (16)

Diastolic 56 (14) 56 (14)

Coma depth (total N, median [IQR])

Blantyre coma score 1704, 4 (2–5) 1713, 4 (2–5)

Glasgow coma score 1005, 11 (8–15) 999, 11 (8–15)

Comorbidity

Immune compromised (from history)

49 (2%) 45 (2%)

Severe malnutrition 43 (2%) 54 (2%)

(Continues in next column)

Quinine(N=2713)

Artesunate(N=2712)

(Continued from previous column)

Suspected pneumonia 226 (8%) 227 (8%)

Confi rmed by radiograph 29 (13%) 29 (13%)

Clinical sepsis 355 (13%) 302 (11%)

Confi rmed by culture 33 (9%) 32 (11%)

Suspected meningitis 166 (6%) 169 (6%)

Confi rmed meningitis 3 (2%) 6 (4%)

Other signifi cant comorbidities 71 (3%) 80 (3%)

Laboratory assessments

Parasitaemia (parasites per μL; geometric mean, range)

49 110 (0–1 858 880)

47 922 (0–1 494 640)

Sodium (mmol/L) 132 (6·5) 131 (6·5)

Potassium (mmol/L) 4·1 (0·9) 4·1 (0·9)

Chloride (mmol/L) 105 (10) 105 (10)

Blood urea nitrogen (mmol/L) 6·1 (4·9) 6·1 (4·6)

Haemoglobin (g/L) 70 (31) 68 (29)

pH 7·36 (0·14) 7·36 (0·14)

PaCO2 (mm Hg) 28·2 (10·1) 27·9 (9·1)

HCO3 (mmol/L) 16·6 (5·7) 16·6 (5·6)

Plasma BE (mmol/L) –8·6 (7·3) –8·5 (7·3)

Anion gap (mmol/L) 17·2 (5·0) 17·0 (4·9)

Data are number (%), median (IQR), or mean (SD), unless otherwise indicated. BE=base excess . PaCO2=partial pressure of carbon dioxide. HCO3=bicarbonate. *See webappendix p 12 for classifi cation of categories. †Depth of coma was assessed either by Blantyre coma score (for preverbal children, n=3417) or Glasgow coma scale (n=2004). ‡Respiratory distress was defi ned as costal indrawing, use of accessory muscles, nasal alar fl aring, deep breathing, or severe tachypnoea. §Severe prostration was defi ned as inability to breastfeed for children younger than 6 months or inability to sit for older children.

Table 1: Baseline characteristics in the two treatment groups

Figure 2: Kaplan-Meier curves comparing survival in African children with severe falciparum malaria treated with either parenteral artesunate or quinineThe numbers in parentheses are the deaths during the indicated time. In eight patients the exact time of death during the night was missing and was estimated as 2359 h.

0

27122713

(158)(186)

(32)(63)

(16)(26)

(13)(10)

(11)(12)

25542527

25222464

25062438

24932428

Number at riskArtesunate

Quinine

0

0·90

Prop

ortio

n st

ill a

live

1·00

1 2

Analysis time (days)

Log-rank p=0·0022

ArtesunateQuinine

3 4

0·95

See Online for webappendix

Correspondence to:Prof N J White, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok 10400, [email protected]

Articles

1650 www.thelancet.com Vol 376 November 13, 2010

aspects of supportive treatment, based on WHO guidelines, were unaff ected by the trial.1,2,9

Although the trial was open label at each site, none of the investigators or trialists, apart from for the trial statistician (TEP), had access to the summaries of treatment allocations. When notes or case record forms were reviewed, all study drug details were removed to preserve masking. All case reviews and laboratory

assessments were done blinded to treatment allocation, which was not revealed until the database was locked at the end of the trial.

ProceduresArtesunate (Guilin Pharmaceutical Factory, Guangxi, China) was given in a dose of 2·4 mg/kg on admission, at 12 h, at 24 h, and thereafter once daily until oral medication

Figure 3: Treatment eff ect in protocol-specifi ed subgroupsThe forest plot shows odds ratios and 95% CIs. The size of the squares is proportional to the size, and therefore weight, of the subgroup. The diamonds show the combined diff erences. The effi cacy of antimalarial pretreatment was classifi ed before study unblinding (webappendix p 12). Hyperparasitaemia means greater than 10% of red cells parasitised. OR=odds ratio. GCS=Glasgow coma scale. BCS=Blantyre coma scale. BE=base excess. *Site mortality classifi ed as low if the site mortality rate was lower than the overall study mortality rate, and high if the site mortality rate was higher than the overall study mortality rate. †Classifi ed according to centre policy (ten sites); classifi ed according to individual data (one site). ‡Decompensated or compensated shock. I2 denotes the percentage of total variation across subgroups resulting from heterogeneity rather than chance, with the p value of signifi cance.

Pretreatment antimalarialNoneIneffectiveEffectiveSubtotal (I2=8·9%, p=0·33)

Site mortality*LowHighSubtotal (I2=0·0%, p=0·52)

Route of treatment†IntramuscularIntravenousSubtotal (I2=0·0%, p=0·99)

Coma (GCS <11 or BCS <3)NoYesSubtotal (I2=0·0%, p=0·37)

Anaemia (<50 g/L)NoYesSubtotal (I2=0·0%, p=0·52)

Shock‡NoYesSubtotal (I2=19·4%, p=0·38)

Acidosis (BE ≤8 mmol/L)NoYesSubtotal (I2=23·0%, p=0·25)

Respiratory distress‡NoYesSubtotal (I2=19·9%, p=0·26)

Hyperparasitaemia (>10%)NoYesSubtotal (I2=0·0%, p=0·32)

86/1281 (7%) 38/387 (10%) 94/938 (10%)

65/1183 (5%) 165/1529 (11%)

91/933 (10%) 139/1779 (8%)

70/1832 (4%) 160/880 (18%)

136/1696 (8%) 77/737 (10%)

173/2389 (7%) 57/323 (18%)

49/1275 (4%) 149/1009 (15%)

169/2273 (7%) 61/439(14%)

140/1766 (8%) 56/584 (10%)

125/1270 (10%) 38/371 (10%) 124/959 (13%)

77/1174 (7%) 220/1539 (14%)

126/989 (13%) 171/1724 (10%)

96/1764 (5%) 199/945 (21%)

163/1734 (9%) 93/692 (13%)

231/2374 (10%) 66/339 (19%)

52/1314 (4%) 189/975 (19%)

213/2285 (9%) 84/428 (20%)

167/1779 (9%) 77/573 (13%)

0·66 (0·49–0·88)1·01 (0·62–1·64)0·74 (0·55–0·98)0·74 (0·61–0·89)

0·83 (0·59–1·16)0·72 (0·58–0·90)0·75 (0·63–0·90)

0·75 (0·56–1·00)0·75 (0·59–0·95)0·75 (0·62–0·90)

0·70 (0·51–0·96)0·84 (0·66–1·06)0·78 (0·65–0·95)

0·85 (0·67–1·08)0·74 (0·53–1·03)0·81 (0·66–0·98)

0·72 (0·59–0·89)0·89 (0·59–1·34)0·75 (0·63–0·91)

0·96 (0·64–1·44)0·73 (0·58–0·93)0·79 (0·64–0·97)

0·78 (0·63–0·97)0·61 (0·42–0·89)0·74 (0·61–0·89)

0·84 (0·66–1·06)0·67 (0·47–0·97)0·78 (0·64–0·96)

1·2510·750·50·4

Artesunate

Events/total (%)

Quinine OR (95% CI)

Events/total (%)

Favours artesunate Favours quinine

Articles

www.thelancet.com Vol 376 November 13, 2010 1651

could be taken reliably. The contents of each 60 mg vial were dissolved initially in 1 mL 5% sodium bicarbonate (provided with the drug) and then diluted with 5% dextrose before injection either as a bolus into an indwelling intravenous cannula, or administration by deep intra-muscular injection to the anterior thigh. Quinine di-hydrochloride (Indus Pharma, Karachi, Pakistan) was given in a 20 mg salt per kg loading dose infused over 4 h (in 5–10 mL/kg of 5% dextrose), followed by a 10 mg salt per kg infusion over 2–8 h three times daily until starting oral therapy. For intramuscular treatment the doses were the same as for intravenous treatment; quinine was diluted in normal saline to a concentration of 60 mg/mL, and injected into the anterior thigh. The loading dose was given as a split dose into each thigh.

When the patient was able to take tablets, but after a minimum of 24 h of parenteral treatment, oral artemether-lumefantrine (Coartem, Novartis, Basel, Switzerland) in a full standard dose (1·5/9 mg/kg twice daily for 3 days with milk or fat) was given to complete the treatment.

A 1 mL blood sample was taken for immediate haematocrit and biochemical analyses with the EC8+ card for a handheld blood analyser (i-STAT, Abbott Laboratories, Abbott Park, IL, USA). This device produced an immediate printed paper report with time of day, which was kept with the case record form. Haemoglobin was reported with the i-STAT result or, when not available, calculated from the measured haematocrit (n=146).10 Thick and thin blood smears were prepared for later malaria parasite counting at the Bangkok reference laboratory. In 109 cases no count was available from the reference laboratory, so the parasitaemia reported by the study site was used. Children were discharged from hospital at the discretion of the physician, and a discharge assessment was completed. Children who had not made a full neurological recovery by discharge were followed up at regular intervals for 12 months or until full recovery. Full neurological assessments were completed at every visit. Training in neurological assessment was provided to all sites by a specialist paediatric neurologist (MO) to ensure uniformity.

Trial sites were monitored regularly by Family Health International, Nairobi, Kenya. Investigators provided reports every 2 weeks to the coordinating centre in Bangkok and met every year to review study progress. Drug content and quality were checked in ampoules taken randomly from the purchase lots (webappendix p 7). The data and safety monitoring committee made three interim analyses during the trial. Adequacy of random-isation was assessed by checking by centre that random-isation was balanced according to baseline variables, and by allocation time to assess whether the expected random variations were observed.

Study outcomesThe analysis was undertaken according to a prespecifi ed analytical plan. The primary outcome measure was in-

hospital mortality compared between treatments on an intention-to-treat basis. Secondary outcome measures were the incidence of severe neurological complications

Figure 4: Neurological sequelae at discharge and after 28 days (range 3–8 weeks) in children with severe falciparum malaria *Some patients had severe impairment in more than one domain.

191 patients with possible neurological deficits atdischarge (110 artesunate, 81 quinine)

21 excluded (11 artesunate, 10 quinine)4 unspecified behavioural problems9 no neurological problem confirmed4 pre-existing neurological problems4 mild motor impairment in young

children <18 months

170 patients with neurological sequelae at discharge(99 artesunate, 71 quinine)

41 lost to follow-up at day 28 (23 artesunate,18 quinine)

68 sequelae resolved by day 28 (42 artesunate,26 quinine)

61 patients with neurological sequelae at day 28(34 artesunate, 27 quinine)

10 mild neurologicalsequelae at day 28(8 artesunate, 2 quinine)

8 moderate neurologicalsequelae at day 28(3 artesunate, 5 quinine)

43 severe neurological sequelae at day 28*(23 artesunate, 20 quinine; 30 severe motor impairment, 14 cortical blindness, 9 severe speech or hearing impairment)

Quinine (n/N, %) Artesunate (n/N, %) OR (95% CI) p value

Mortality, ITT analysis 297/2713 (10·9%) 230/2712 (8·5%) 0·75 (0·63–0·90) 0·0022

Mortality, per-protocol analysis 260/2552 (10·2%) 208/2563 (8·1%) 0·78 (0·64–0·94) 0·0099

Death or sequelae at 28 days 316/2695 (11·7%) 253/2689 (9·4%) 0·78 (0·65–0·93) 0·0056

Malaria-attributable mortality* 288/2704 (10·7%) 223/2705 (8·2%) 0·75 (0·63–0·91) 0·0025

Mortality in strictly defi ned severe malaria†

291/2338 (12·4%) 226/2280 (9·9%) 0·77 (0·64–0·93) 0·0055

Case fatality in HIV-positive children‡

19/61 (31%) 16/64 (25%) 0·74 (0·33–1·62) 0·45

Development of coma§ 91/1768 (5·1%) 65/1832 (3·5%) 0·69 (0·49–0·95) 0·0231

Deterioration of coma score 208/2713 (7·7%) 166/2712 (6·1%) 0·78 (0·64–0·97) 0·0245

Convulsions developing or persisting >6 h after admission

273/2713 (10·1%) 224/2712 (8·3%) 0·80 (0·66–0·97) 0·0199

Hypoglycaemia 75/2713 (2·8%) 48/2712 (1·8%) 0·63 (0·43–0·91) 0·0134

Severe anaemia (<50 g/L) after admission§

98/1734 (5·7%) 78/1696 (4·6%) 0·81 (0·59–1·11) 0·18

Blackwater fever§ 18/2597 (0·7%) 30/2591 (1·2%) 1·69 (0·94–3·05) 0·076

ITT=intention to treat. *The likelihood that malaria contributed to or directly caused the death was assessed by an independent endpoint review committee blinded to the treatment allocation. †As defi ned in panel 1. ‡HIV status was assessed only in Beira, Muheza, and Kilifi (n=2095). §Development of coma, anaemia, and blackwater fever was assessed only in patients without these disorders on admission.

Table 2: Mortality and complications according to treatment group

Articles

1652 www.thelancet.com Vol 376 November 13, 2010

(assessed at 28 days, range 3–8 weeks) and a combined outcome measure of death and severe persistent neurological sequelae. Initially, neurological outcomes were assessed only at discharge from hospital, but this procedure led to substantial overestimation of neuro-logical defi cit, especially in young children. The protocol was therefore changed in April, 2007, (after 11% of patients had been enrolled) so that children who had not yet fully recovered at discharge were assessed 28 days after enrolment, and active follow-up was instituted. Outcome measures were also assessed per protocol, excluding patients not fulfi lling the entry criteria, those with a negative or missing admission blood slide for P falciparum, those dying before receiving the study drug, and those missing a study drug dose on the fi rst day of treatment. Subgroup analyses included stratifi cation into two predefi ned groups: those who fulfi lled the criteria for severe malaria1,2,11 and those who did not. In sites at which testing was approved, the HIV status of participants was assessed after obtaining a separate informed consent, and the outcomes in HIV-1 infected patients with malaria were compared according to treatment allocation.

Two committees were formed to provide independent expert assessment and classifi cation of outcomes before study unblinding. A neurological outcome committee, comprising two experienced paediatricians (both with neurological expertise and experience of working in Africa) and one clinical malariologist, graded neurological sequelae. These sequelae were assessed in four domains: motor function, visual function, hearing, and speech, and also whether the child had developed epilepsy. Sequelae persisting at follow-up were graded as mild, moderate, or severe in each domain according to the extent of functional impairment (webappendix p 5). The overall grade was combined. If there were sequelae in more than one functional domain the grade was increased. The relation of sequelae to severe malaria was assessed taking into account any pre-existing comorbidity. A separate endpoint review committee, comprising one paediatrician with malaria experience and one clinical malariologist, identifi ed cases in which a pathological change other than malaria or its acute complications was likely to be the main cause of death.

Statistical analysisInclusion of more than 5306 children with severe malaria was needed to show, with 80% power and 95% confi dence, a 25% reduction in mortality from 8% to 6%. This calculation was based on an estimated severe malaria mortality of 16% with about half the enrolled patients anticipated to fulfi l the criteria for severe malaria. Patient data and outcomes were provided to an independent data and safety monitoring committee, who reviewed the trial yearly.

For binary outcomes, the odds ratios (ORs) between treatment groups, stratifi ed by study site, were estimated by the Mantel-Haenszel method. Heterogeneity between sites was examined with the Breslow-Day test. In Rwanda, the two small sites run by the same investigators were pooled. Time to event outcomes were compared with the log-rank test, and hazard ratios (HRs) were estimated by a Cox proportional hazard model, stratifi ed by site. Statistical analyses were done with Stata (version 11.1).

This trial is registered, number ISRCTN50258054.

Role of the funding sourceThe sponsor of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report. The corresponding author had full access to all the data in the study and had fi nal responsibility for the decision to submit for publication.

ResultsFigure 1 shows the trial profi le. We recorded no major diff erences in baseline characteristics between the two treatment groups (table 1). 5425 patients were recruited (fi gure 1), of whom 527 (9·7%) died. 230 of 2712 (8·5%) patients given artesunate died versus 297 of 2713 (10·9%) given quinine (relative risk 0·78, 95% CI 0·66–0·91; OR 0·75, 0·63–0·90, in favour of artesunate; p=0·0022). This represents a relative reduction in mortality of 22·5% (95% CI 8·1–36·9%) and corresponds to an overall number needed to treat to prevent one death of 41 (95% CI 25–112). We recorded no heterogeneity between study sites (p=0·99). Eight patients (fi ve artesunate, three quinine) were taken from hospital against advice and could not be followed up further. They were censored in the survival analysis at the time of discharge. The survival analysis (fi gure 2) for overall mortality during admission by antimalarial treatment gives the same result as the Mantel-Haenszel analysis (HR stratifi ed by study site 0·76, 95% CI 0·64–0·91; p=0·0022). Mantel-Haenszel analysis of the predefi ned subgroups showed no evidence of any diff erences in odds ratios between subgroups (fi gure 3), and these results were confi rmed by Cox regression (data not shown).

The per-protocol analysis excluded patients who died rapidly before receiving antimalarial treatment (six artesunate, 22 quinine; p=0·0023), patients with incomplete initial antimalarial treatment with the study drug, and those with negative or missing blood slides for

Quinine (median, IQR)

N Artesunate (median, IQR)

N HR (95% CI) p value

Time to discharge (days) 3·0 (2·0–5·0) 2412 3·0 (2·0–5·0) 2478 1·04 (0·99–1·10) 0·059

Time to eat (h) 12 (2–24) 2269 9 (0–24) 2358 0·99 (0·93–1·06) 0·74

Time to sit unsupported (h) 22 (6–44) 2312 18 (6–42) 2373 1·02 (0·95–1·08) 0·60

Time to localise pain (h)* 12 (6–24) 726 12 (6–24) 698 0·87 (0·78–0·98) 0·0093

Time to speak (h)* 18 (11–36) 695 20 (8–42) 664 0·88 (0·79–0·99) 0·016

*Time to localise pain and time to speak was assessed only for surviving patients with coma on admission (Blantyre coma scale <3 or Glasgow coma scale <11).

Table 3: Recovery times in surviving patients according to treatment group

Articles

www.thelancet.com Vol 376 November 13, 2010 1653

P falciparum (fi gure 1), but these omissions did not substantially aff ect the result (table 2). The endpoint review committee identifi ed 16 children (seven artesunate, nine quinine) in whom death was unlikely to be related to severe malaria. Omission of these cases from the analysis also had no eff ect on the magnitude of the survival benefi t with artesunate (table 2 and webappendix p 3). In 4618 children fulfi lling strict criteria for severe malaria (panel 1), mortality was 9·9% (226/2280) with artesunate and 12·4% (291/2338) with quinine (OR 0·77, 95% CI 0·64–0·93; p=0·0055). Eight fatal cases (fi ve quinine, three artesunate) did not have an accurate time of death recorded. In the remainder, 345 of 519 deaths occurred within the fi rst 24 h after admission, of which 158 of 2709 (5·8%) were assigned to artesunate treatment and 187 of 2708 (6·9%) to quinine treatment (OR 0·84, 95% CI 0·67–1·04, p=0·109). Of the 174 of 519 deaths that occurred more than 24 h after admission, 69 of 2551 (2·7%) patients were treated with artesunate and 105 of 2521 (4·2%) with quinine (OR 0·63, 0·47–0·87, p=0·004). HIV serology was assessed routinely in Beira, Muheza, and Kilifi . Of 2095 patients tested, 125 (6%) were positive (64 artesunate, 61 quinine). Mortality in these patients was high (table 2).

497 children (224 artesunate, 273 quinine) had convulsions that either developed after admission, irrespective of duration, or were present on admission and persisted for more than 6 h, and 156 patients (65 artesunate, 91 quinine) developed coma or had a deterioration of their coma score after starting anti malarial treatment (table 2). The development of coma, deterioration in coma score, and convulsions all occurred more frequently in patients who received quinine than in those who received artesunate (table 2). In the 4898 survivors, 170 (99 artesunate, 71 quinine) had not yet made full neurological recoveries at discharge (fi gure 4). Of these patients, 129 (76 artesunate, 53 quinine) were followed up between 3 and 8 weeks after

enrolment. At this fi rst follow-up assessment 68 (53%) had recovered fully, 18 (14%; 11 artesunate, seven quinine) were mildly or moderately impaired, and 43 (33%; 20 quinine, 23 artesunate) had severe neurological defi cits. The overall incidence of any persistent neurological sequelae in assessed survivors at 28 days after cerebral malaria was 3·2% (24/706 artesunate, 23/737 quinine) and of severe neurological sequelae was 2·3% (17/706 artesunate, 17/737 quinine). We recorded no diff erence between the treatment groups. Of the 14 patients with any neurological sequelae who did not have cerebral malaria initially (ten artesunate, four quinine), seven had multiple convulsions (three quinine, four artesunate) and all had severe prostration on admission.

We detected no severe adverse eff ects that could be attributed directly to drug toxicity. Although one patient treated with artesunate developed a mild urticarial rash, no severe type 1 hypersensitivity reactions were recorded. Another patient treated with artesunate developed peripheral gangrene of toes and fi ngers, which was attributed to the disease and not the drug. One patient given quinine developed severe stridor after administration of ampicillin, and died. This death was attributed to ampicillin rather than quinine. Hypoglycaemia after starting antimalarial treatment was signifi cantly less frequent in patients who received artesunate than in those who received quinine (table 2). Blackwater fever was rare in both groups (table 2).

Blood transfusions were given to 1487/2712 (55%) patients assigned to artesunate and 1495/2713 (55%) assigned to quinine. A fl uid bolus at the start of treatment was given to 589/2712 (22%) patients assigned to artesunate and 596/2713 (22%) assigned to quinine. 3259 (60%) children received antibiotics (1606 artesunate, 1653 quinine; p=0·20).

In cerebral malaria survivors, the time from randomisation until the child was able to localise a

Figure 5: Meta-analysis of all randomised controlled trials that have compared mortality of severe malaria in patients treated with parenteral artesunate versus parenteral quinine12–16

The solid vertical line represents equality of the two groups; the dashed line is the overall treatment diff erence. The horizontal lines and the width of the diamonds show the CIs for the odds ratios. The size of the squares is proportional to the size, and therefore weight, of the trial. OR=odds ratio. *99% CIs for totals.

AfricaAQUAMAT 2010Sudan (2010)16

Subtotal (test for heterogeneity: χ2=0·06, df=1, p=0·81)

AsiaSEAQUAMAT (2005)7 Thailand (2003)15

Vietnam (1997)14

Vietnam (1992)13

Burma (1992)12

Subtotal (test for heterogeneity: χ2=1·80, df=4, p=0·77)

Overall (test for heterogeneity: χ2=4·51, df=6, p=0·61)Heterogeneity between continents: χ2=2·65, df=1, p=0·10

297/2713 (10·9%) 2/33 (6·1%)

164/731 (22·4%) 12/54 (22·2%) 5/35 (14·3%) 8/30 (26·7%) 23/67 (34·3%)

230/2712 (8·5%) 1/33 (3%)

107/730 (14·7%) 7/59 (11·9%) 4/37 (10·8%) 5/31 (16·1%) 2/24 (8·3%)

0·75 (0·63–0·90); 0·0020·48 (0·01–9·85); 0·560·75 (0·59–0·95); 0·002

0·60 (0·45–0·79); 0·00020·47 (0·14–1·44); 0·140·73 (0·13–3·75); 0·660·53 (0·12–2·17); 0·320·17 (0·02–0·83); 0·020·58 (0·41–0·81); 0·00005

0·69 (0·57–0·84); <0·00001

10·1 0·3 0·7 2 3 4

Artesunate Quinine OR (95% CI)*; p value

Favours artesunate Favours quinine

Articles

1654 www.thelancet.com Vol 376 November 13, 2010

painful stimulus or was able to speak was slightly longer overall in patients treated with artesunate than in those given quinine when compared by survival analysis (table 3). The times to eat or to sit unsupported did not diff er between treatment groups (table 3).

The content of artesunate in all ampoules tested was within the pharmaceutical content limits of 10% of the stated active ingredient. All tested vials of quinine contained between 105% and 112% of the stated content (webappendix p 7).

In a meta-analysis of all severe malaria trials that have compared survival after parenteral artesunate with that after parenteral quinine, the overall OR was 0·69 (0·57–0·84; p<0·00001) in favour of artesunate (fi gure 5). We noted no signifi cant heterogeneity between the results generated in Africa and Asia (fi gure 5).

DiscussionThis large multicentre trial shows that artesunate substantially reduces the overall mortality of African children diagnosed with severe malaria. We recorded little heterogeneity between treatment centres in the benefi t associated with artesunate, suggesting that the fi ndings are robust. Before this trial, concerns had been raised that artesunate might not be better than quinine in African children, whereas it clearly was in Asian patients. The concerns arose because of perceived diff erences in pathology and the prevalence of more quinine-susceptible malaria parasites, and because in the earlier SEAQUAMAT trial undertaken in Asia,7 the survival curves did not separate clearly until 48 h after starting treatment, whereas most deaths in African children occurred before this time. Fortunately these concerns were not substantiated. More than 7000 severely ill patients have now been included in randomised comparisons of parenteral artesunate and quinine (fi gure 5). Compared

with parenteral quinine, artesunate reduced the mortality of severe malaria in African children by 22·5%, and in Asian patients by 38·6%.7 These studies together comprise nearly 80% of all patients ever enrolled in randomised controlled trials of patients admitted with severe malaria, and they provide defi nitive evidence that artesunate is the most eff ective available treatment for severe malaria, and that it is safe (panel 2).

This multicentre trial was open label because the substantial diff erences in the parenteral formulations of the two drugs prevented adequate concealment. Although individual trial site investigators knew the individual treatment allocations, all the other clinical and laboratory investigators were masked to the parenteral treatments given. The similarity of results across the diff erent trial sites suggests that signifi cant bias was unlikely. The great strength of this trial is its unprecedented size, its consistency both internally and with previous studies, and therefore the confi dence that health-care workers throughout the tropical world can have in its result.

Artesunate prevented death from severe malaria, but not at the expense of an increase in neurological sequelae. Indeed, the overall incidence of confi rmed persistent severe sequelae after severe malaria was low.17 Initially the assessment of neurological defi cit was made at discharge from hospital, but many children, especially in the younger age group, had not made a full neurological recovery at discharge. Subsequently, assessments were made 4 weeks later, by which time about half the apparent defi cits had resolved fully.

This life-saving benefi t of artesunate compared with quinine in severe malaria has to derive from its greater intrinsic parasiticidal activity. The principal pharma-codynamic advantage of artesunate is that it has a much broader stage-specifi city of action than does quinine.18 The artemisinins kill circulating ring-stage parasites before they can mature,19,20 which reduces sequestration of infected erythrocytes in the venules and capillaries of vital organs and thereby prevents potentially lethal microvascular obstruction.21–23 The large and consistent reduction in mortality associated with artesunate, and the consistent fi nding that mortality reduction is greatest in hyperparasitaemia,7 lends support to the central quantitative role of parasitised erythrocyte sequestration in the pathology of malaria.

The benefi ts of parenteral artesunate compared with quinine were greater than the benefi ts of intramuscular artemether reported in previous trials.6,7 In a double-blind trial24 comparing artesunate and artemether in 370 Vietnamese adults with severe falciparum malaria, 13 patients died in the artesunate group (7%) and 24 in the artemether group (13%). Taken together, these diff erent study results suggest that the live-saving benefi t compared with quinine provided by artesunate is roughly twice that provided by artemether. Artesunate and artemether have similar pharmacodynamic properties in vitro, so the most likely explanation for their diff erent

Panel 2: Research in context

Systematic reviewWe searched Medline (from January, 1966, to September, 2010) and Embase (from January, 1980, to September, 2010) for randomised controlled trials. Search terms used were: “malaria”, “cerebral malaria”, “severe malaria”, “complicated malaria”, “malaria falciparum”, “quinine”, and “cinchona alkaloids” (MeSH/EMTREE terms); and “artemisinin” and “artesunate”. We searched for randomised controlled trials that compared parenteral artesunate with quinine for treatment of severe malaria. We identifi ed six randomised controlled trials,7,12–16 which we have included in the meta-analysis presented in fi gure 5.

InterpretationTogether these trials provide substantial evidence of the life-saving benefi t of artesunate compared with quinine in the treatment of severe falciparum malaria in all age groups worldwide.

Articles

www.thelancet.com Vol 376 November 13, 2010 1655

effi cacies in vivo is the substantial diff erence in their pharmacokinetic properties. Parenteral artemether is an oil-based formulation given only by intramuscular injection. Absorption is slow and erratic, whereas the water-soluble hemisuccinate artesunate is absorbed rapidly and reliably after intramuscular injection and can be given intravenously.25–30 Thus the pharmacodynamic advantage of artemether over quinine might have been off set by its poor absorption kinetics. In summary, in the treatment of severe falciparum malaria, it seems that artesunate is better than artemether, which in turn is better than quinine.

The benefi ts of artesunate (and artemether) compared with quinine were greater in patients from southeast Asia than in African children,6,7 although the studies were not large enough to show this diff erence reliably. There are several possible explanations for this fi nding. African children usually have some background immunity that assists the therapeutic response, and in particular accelerates circulating parasite clearance.31 This eff ect hastens recovery and could reduce the therapeutic advantage of artesunate. The greater quinine susceptibility of P falciparum in Africa is often proposed as contributing to improved therapeutic responses, but the diff erences are not large, and are unlikely to have a major eff ect in vivo. Incorrect diagnosis is likely to be a major contributory factor; severe malaria is overdiagnosed in African children, and sepsis is underdiagnosed.32 Febrile sick children with positive malaria blood smears are usually diagnosed initially as having severe malaria, but the specifi city of a positive blood smear is poor in settings in which a high proportion of all children are parasitaemic. Septicaemia and pneumonia are especially diffi cult to diff erentiate clinically from severe malaria. Sepsis and severe malaria also commonly coexist in African children.32 Prompt and appropriate treatment of sepsis should further reduce the mortality of severely ill children with malaria parasitaemia.

Parenteral artesunate is simple to administer, is safe, and reduces mortality substantially compared with quinine. No serious adverse eff ects were identifi ed in this large study or in previous large studies.7,24,33 By contrast, intramuscular quinine is locally toxic4 (because of its acidity), and intravenous quinine needs a carefully rate-controlled infusion and continuous or three times daily administration to avoid dangerous hypotension.9 Importantly, quinine is associated with potentially serious hypoglycaemia.1,2,5 In this trial, 22 children died before receiving quinine compared with only six who died before receiving artesunate. This fi nding is probably indicative of the diffi culties in administering parenteral quinine promptly and safely. Any delay in treating severe infection will increase mortality.33 The ease and safety of parenteral artesunate are important practical advantages. Artesunate is more expensive to buy, but quinine is more expensive to administer. A major factor restricting the deployment of artesunate has been unavailability of a

product satisfying international good manufacturing standards. The most widely used product, assessed in this study, does not yet have this certifi cation, which has prevented deployment in some countries.34–39 This barrier must be overcome speedily so that parenteral artesunate can be deployed in malaria-endemic areas to save lives.

Artesunate should now become the treatment of choice for severe malaria for children and adults worldwide. Malaria causes an estimated 800 000 deaths every year in African children.40 Severe malaria is often the most common admission diagnosis in febrile children, so a change in treatment policy from quinine to artesunate has the potential to save thousands of children’s lives every year. If 4 million African children with severe malaria every year were to receive prompt treatment with parenteral artesunate instead of quinine, and the benefi ts were similar to those recorded in this trial, then approximately 100 000 lives might be saved per year.

ContributorsThe coordinating committee designed the study. All investigators in the

trial sites undertook the trial, with support from the team in Bangkok.

All investigators and the coordinating committee reviewed and

discussed the trial results. The writing committee did the data analysis

and prepared the report.

The AQUAMAT trial group Writing committee: Arjen M Dondorp, Caterina I Fanello,

Ilse C E Hendriksen, Tim E Peto, Lorenz von Seidlein, Nicholas P J Day,

Nicholas J White.

Clinical trialists: Hospital Central da Beira, Beira, Mozambique

Ermelinda Gomes (principal investigator [PI]), Amir Seni,

Kajal D Chhaganlal, Ilse C E Hendriksen, Alínia José Pedro,

César D R Jarach, Olivier Wingi, Ester I Fernando, Mara Zambruni,

Zacarias Raimundo, Carlos J de Oliveira, Arjen Dondorp,

Francisco Songane, Avertino Barreto, Marcelino Lucas; MRC laboratories and Royal Victoria Teaching Hospital, Banjul, The Gambia

Kalifa Bojang PI, Rasaq Olaosebikan, Nkechinyere Anunobi,

Tumani Corrah, David Conway, Sarah Rowland-Jones, Francis Akor,

Joseph Okebe, Muminatou Jallow, Osaretin Chimah,

Abubakar Ismaela, Mamkumba Sanneh, Saidykhan Mamodou,

Fatou Jammeh, Janet Fullah, Abdou Bah, Horija Saine; Komfo Anokye Hospital, Kumasi, Ghana Tsiri Agbenyega PI, Samuel Blay Nguah,

Jennifer Evans, Daniel Ansong, Justice Sylverken, Alex O Akoto,

Larko D Owusu, David Sambian, Michael Ntim; Kilifi District General Hospital, Kilifi , Kenya Kathryn Maitland PI (and Wellcome Trust Centre

for Clinical Tropical Medicine, Faculty of Medicine, Imperial College,

London, UK), Esther Kivaya, Mohammed Abubakar, Ayub Mpoya,

Charles Newton (and Neurosciences Unit, The Wolfson Centre,

Institute of Child Health, University College of London, UK),

Samson Gwer, Samuel Akech, Michael Kazungu, Michael Kihara,

Molline Timbwa, Mwanamvua Boga; Magunga District Hospital, NIMR-Korogwe Research Laboratory, Tanga, Tanzania Samwel Gesase PI,

Catherine Kahabuka, Martha Lemnge, Lorenz von Seidlein,

Aileen Barongo, Anangisye Malabeja, Omary Abdul, Zacharia Savael,

Gustav Tilaus, Laura Chuwa, Stella Kabela; Teule Designated District Hospital, Muheza, Tanzania Behzad Nadjm, Jacqueline Deen,

Hugh Reyburn, Christopher Whitty, Ilse C E Hendriksen, Amani

Shao, Selemani Mtunguja, Marwa Mwikwabe, Walli Msuya,

Christina Kiemi, Edward Mtili, Lorenz von Seidlein, Emmanuel Swai,

Ben Amos, Rajabu Malahiyo, Regina Malugu, Michael Mnongea,

Stella Emmanuel, Nzitu J Msengi, Faraja M Mtimbo, Juma Kimera;

NIMR-Amani Centre, Tanga, Tanzania George Mtove; Rwamagana Hospital and Nyanza Hospital, Rwanda and Malaria Control Program MoH Kigali Corine Karema PI, Noella Umulisa, Aline Uwimana,

Josephine Ngalula Mulumba, Aimee Uwase, Devotha Rugandura,

Claude Kawite, Deogratias Ruhangaza, Jean Claude Ndagijimana,

Josephine Mukeshiyaremye, Antoinette Kayitesi,

Articles

1656 www.thelancet.com Vol 376 November 13, 2010

Violette Mukamugenzi, Ntwali Ndizeye, Tharcisse Munyaneza;

University of Ilorin Teaching Hospital, Ilorin, Nigeria

Olugbenga A Mokuolu PI, Olanrewaju T Adedoyin,

Wahab B R Johnson, Ayodele Ojuawo, Samuel K Ernest,

Omotayo O Adesiyun, Aishatu A Abdulkarim,

Abdulrasheed O Adegboye, Ashau O Timi-Oladipo, Bukola R Kelani,

Oyekunle Ogunkanbi, Abdulrahreem Yusuf, Oluyemisi Adegboye,

Olabode T Oyewale, Olusegun Oladele, Alice Jacob, Oluseun Faniyi;

Epicentre Research Base Mbarara and Mbarara University Teaching Hospital, Mbarara, Uganda Juliet Mwanga-Amumpaire PI,

Margaret Nansumba, Patrice Piola, Pierre Debeaudrap, Yap Boum II,

Mehul Dhorda, Eleanor Turyakira, Elias Kumbakumba,

Eunice Nyesigire, Peters Kalubi, Santorino Data, Naome Natukunda,

Antoney Ssegane, Nyehangane Dan, Benon Tumwebaze,

Benson Twinomujuni, James Kinyatta, Suzan Esther Asaasira,

Jacqueline Tumuhairwe, Sanyu Scovia, Primativa Kyohairwe,

Shiela Mbabazi, Harriet Adrama, Thaddeus Turuho,

Sulaiman Muwanga, John Bosco Ariyo, Peace Magezi, Agatha

Twebaze; Kinshasa School of Public Health, Kingasani Research Centre, Kinshasa, Democratic Republic of the Congo Antoinette K Tshefu PI,

Marie A Onyamboko, Jacques K Zandibeni, Elyse K Katokale,

Charlie N Kabedi, Adolphine B Nkuadiolandu, Raoul N Mpoyi,

Marcelline M Sakina, Crispin K Fela, Jacqueline A Omari,

Odile K Muniaka, Betty D Omba, Antoinette I Boyanga,

Elisee K Biyela, Benjamin B Badjanga, Yvonne B Kambashi.

Clinical, laboratory, statistical and logistic support: Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand Arjen M Dondorp, Tharisara Sakulthaew,

Caterina I Fanello, Wirichada Pan Ngum, Kasia Stepniewska,

Kamolrat Silamut, Ilse C E Hendriksen, Nuttapol Panachuenwongsakul,

Montri Rijaibun, Niklas Lindegardh, Warunee Hanpithakpong,

Forradee Nuchsongsin, Benjamas Intarabut, Ketsanee Srinamon,

Somporn Saiwaew, Kesinee Chotivanich, Nicholas J White,

Nicholas P J Day.

Coordinating committee: Caterina I Fanello (study coordinator),

Arjen M Dondorp, Ilse C E Hendriksen, Nicholas P J Day,

Lorenz von Seidlein, Nicholas J White (chair).

Trial steering committee: Kalifa Bojang, Kathryn Maitland,

Arjen M Dondorp, Nicholas P J Day, Nicholas J White,

Brian Greenwood (chair).

Neurological outcome review committee: Martina Oneko, Bridget Wills,

Nicholas White.

Endpoint review committee: Charles J Woodrow, Delia Bethell.

Trial statistician: Tim E Peto.

Data and Safety Monitoring Committee: David Lalloo (chair),

Sarah Walker, Kojo Koram, Malcolm Molyneux, Grace Malenga.

Confl icts of interest NJW is co-chairman and OAM is a member of the WHO antimalarial

treatment guidelines committee. JD was Temporary Adviser for WHO

Model List of Essential Medicines for Children in 2007–08. All other

authors declare that they have no confl icts of interest.

AcknowledgmentsTEP is funded by the National Institute for Health Research Oxford

Biomedical Research Centre. This trial was supported by

grants 076908 and 082541 from the Wellcome Trust, and was

coordinated as part of the Wellcome Trust Mahidol University Oxford

Tropical Medicine Research Programme funded by the Wellcome

Trust of Great Britain. We thank all the patients and their families, the

directors and staff of the trial hospitals and the Bangkok coordinating

centre, and the many doctors, research nurses, and research assistants

for their help and support; the ministries of health and institutional

authorities for their support; Loice Magaria and Ghiorghis Belai for

their assistance and support; Novartis for providing artemether-

lumefantrine for several of the study sites; Marja Schilstra for help

with database construction; Epicentre (Philippe Guerin and

Elizabeth Ashley) for assistance in setting up the study in Uganda;

and Richard Peto for advice.

References1 WHO. Severe and complicated malaria. Trans R Soc Trop Med Hyg

1990; 84 (suppl 2): 1–65.

2 WHO. Severe and complicated malaria. Trans R Soc Trop Med Hyg 2000; 94 (suppl 1): 1–90.

3 White NJ. The treatment of malaria. N Engl J Med 1996; 335: 800–06.

4 Yen LM, Dao LM, Day NP, et al. Role of quinine in the high mortality of intramuscular injection tetanus. Lancet 1994; 344: 786–77.

5 White NJ, Warrell DA, Chanthavanich P, et al. Severe hypoglycemia and hyperinsulinemia in falciparum malaria. N Engl J Med 1983; 309: 61–66.

6 Artemether-Quinine Meta-analysis Study Group. A meta-analysis using individual patient data of trials comparing artemether with quinine in the treatment of severe falciparum malaria. Trans R Soc Trop Med Hyg 2001; 95: 637–50.

7 Dondorp A, Nosten F, Stepniewska K, Day N, White NJ, for the South East Asian Quinine Artesunate Malaria Trial (SEAQUAMAT) group. Artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet 2005; 366: 717–25.

8 Lubell Y, Yeung S, Dondorp AM, et al. Cost-eff ectiveness of artesunate for the treatment of severe malaria. Trop Med Int Health 2009; 14: 332–37.

9 WHO. The treatment of malaria, 2nd edn. Geneva: World Health Organization, 2010.

10 Lee SJ, Stepniewska K, Anstey N, et al. The relationship between the haemoglobin concentration and the haematocrit in Plasmodium falciparum malaria. Malar J 2008; 7: 149.

11 Hien TT, Day NPJ, Phu NH, et al. A controlled trial of artemether or quinine in Vietnamese adults with severe falciparum malaria. N Engl J Med 1996; 335: 76–83.

12 Win K, Than M, Thwe Y. Comparison of combinations of parenteral artemisinin derivatives plus oral mefl oquine with intravenous quinine plus oral tetracycline for treating cerebral malaria. Bull World Health Organ 1992; 70: 777–82.

13 Hien TT, Arnold K, Vinh H, et al Comparison of artemisinin suppositories with intravenous artesunate and intravenous quinine in the treatment of cerebral malaria. Trans R Soc Trop Med Hyg 1992; 86: 582–83.

14 Cao XT, Bethell DB, Pham TP, et al. Comparison of artemisinin suppositories, intramuscular artesunate and intravenous quinine for the treatment of severe childhood malaria. Trans R Soc Trop Med Hyg 1997; 91: 335–42.

15 Newton P, Angus BJ, Chierakul W, et al. A randomised comparison of intravenous artesunate or quinine in the treatment of severe falciparum malaria. Clin Infect Dis 2003; 37: 7–16.

16 Eltahir HG, Omer AA, Mohamed AA, Adam I. Comparison of artesunate and quinine in the treatment of Sudanese children with severe Plasmodium falciparum malaria. Trans R Soc Trop Med Hyg 2010; 104: 684–86.

17 Idro R, Jenkins NE, Newton CR. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol 2005; 4: 827–40.

18 ter Kuile F, White NJ, Holloway P, Pasvol G, Krishna S. Plasmodium falciparum: in-vitro studies of the pharmacodynamic properties of drugs used for the treatment of severe malaria. Exp Parasitol 1993; 76: 86–95.

19 Gravenor MB, van Hensbroek MB, Kwiatkowski D. Estimating sequestered parasite population dynamics in cerebral malaria. Proc Natl Acad Sci USA 1998; 95: 7620–24.

20 Murphy S, Watkins WM, Bray PG, et al. Parasite viability during treatment of severe falciparum malaria: diff erential eff ects of artemether and quinine. Am J Trop Med Hyg 1995; 53: 303–05.

21 Chotivanich K, Udomsangpetch R, McGready R, et al. The central role of the spleen in malaria parasite clearance. J Infect Dis 2002; 185: 1538–41.

22 Udomsangpetch R, Pipitaporn B, Krishna S, et al. Antimalarial drugs reduce cytoadherence and rosetting of Plasmodium falciparum. J Infect Dis 1996; 173: 691–98.

23 Dondorp AM, Ince C, Charunwatthana P, et al. Direct in vivo assessment of microcirculatory dysfunction in severe falciparum malaria. J Infect Dis 2008; 197: 79–84.

24 Phu NH, Tuan PQ, Day NPJ, et al. Randomized controlled trial of artesunate or artemether in Vietnamese adults with severe falciparum malaria. Malar J 2010; 9: e97.

25 Murphy SA, Mberu E, Muhia D, et al. The disposition of intramuscular artemether in children with cerebral malaria; a preliminary study. Trans R Soc Trop Med Hyg 1997; 91: 331–34.

Articles

www.thelancet.com Vol 376 November 13, 2010 1657

26 Silamut K, Newton PN, Teja-Isavadharm P, et al. Artemether bioavailability after oral or intramuscular administration in uncomplicated falciparum malaria. Antimicrob Agents Chemother 2003; 47: 3795–98.

27 Mithwani S, Aarons L, Kokwaro GO, et al. Population pharmacokinetics of artemether and dihydroartemisinin following single intramuscular dosing of artemether in African children with severe falciparum malaria. Br J Clin Pharmacol 2004; 57: 146–52.

28 Hien TT, Davis TM, Chuong LV, et al. Comparative pharmacokinetics of intramuscular artesunate and artemether in patients with severe falciparum malaria. Antimicrob Agents Chemother 2004; 48: 4234–49.

29 Ilett KF, Batty KT, Powell SM, et al. The pharmacokinetic properties of intramuscular artesunate and rectal dihydroartemisinin in uncomplicated falciparum malaria. Br J Clin Pharmacol 2002; 53: 23–30.

30 Nealon C, Dzeing A, Muller-Romer U, et al. Intramuscular bioavailability and clinical effi cacy of artesunate in Gabonese children with severe malaria. Antimicrob Agents Chemother 2002; 6: 933–99.

31 Stepniewska K, Ashley E, Lee SJ, et al. In vivo parasitological measures of artemisinin susceptibility. J Infect Dis 2010; 201: 570–79.

32 Gwer S, Newton CR, Berkley JA. Over-diagnosis and co-morbidity of severe malaria in African children: a guide for clinicians. Am J Trop Med Hyg 2007; 77 (suppl): 6–13.

33 Gomes MF, Faiz MA, Gyapong JO, et al, for the Study 13 Research Group. Pre-referral rectal artesunate to prevent death and disability in severe malaria: a placebo-controlled trial. Lancet 2009; 373: 557–66.

34 Bartoloni A, Tomasoni L, Bartalesi F, et al. Combined intravenous treatment with artesunate and quinine for severe malaria in Italy. Am J Trop Med Hyg 2010; 83: 274–76.

35 Rosenthal PJ. Artesunate for the treatment of severe falciparum malaria. N Engl J Med 2008; 358: 1829–36.

36 White NJ, Day NP, Dondorp A, Anstey N. UK recommendations for severe malaria are worrying. BMJ 2007; 334: 490.

37 Lalloo DG, Shingadia D, Pasvol G, et al, for the HPA Advisory Committee on Malaria Prevention in UK Travellers. UK malaria treatment guidelines. J Infect 2007; 54: 111–21.

38 Jelinek T. Intravenous artesunate recommended for patients with severe malaria: position statement from TropNetEurop. Euro Surveill 2005; 10: 2841.

39 Anstey NM, Price RN, White NJ. Improving the availability of artesunate for treatment of severe malaria. Med J Aust 2006; 184: 3–4.

40 Black RE, Cousens S, Johnson HL, et al. Child Health Epidemiology Reference Group of WHO and UNICEF. Global, regional, and national causes of child mortality in 2008: a systematic analysis. Lancet 2010; 375: 1969–87.